Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 2058, 2023 04 12.
Article in English | MEDLINE | ID: mdl-37045841

ABSTRACT

WHIM Syndrome is a rare immunodeficiency caused by gain-of-function CXCR4 mutations. Here we report a decrease in bone mineral density in 25% of WHIM patients and bone defects leading to osteoporosis in a WHIM mouse model. Imbalanced bone tissue is observed in mutant mice combining reduced osteoprogenitor cells and increased osteoclast numbers. Mechanistically, impaired CXCR4 desensitization disrupts cell cycle progression and osteogenic commitment of skeletal stromal/stem cells, while increasing their pro-osteoclastogenic capacities. Impaired osteogenic differentiation is evidenced in primary bone marrow stromal cells from WHIM patients. In mice, chronic treatment with the CXCR4 antagonist AMD3100 normalizes in vitro osteogenic fate of mutant skeletal stromal/stem cells and reverses in vivo the loss of skeletal cells, demonstrating that proper CXCR4 desensitization is required for the osteogenic specification of skeletal stromal/stem cells. Our study provides mechanistic insights into how CXCR4 signaling regulates the osteogenic fate of skeletal cells and the balance between bone formation and resorption.


Subject(s)
Immunologic Deficiency Syndromes , Osteoporosis , Primary Immunodeficiency Diseases , Receptors, CXCR4 , Animals , Mice , Immunologic Deficiency Syndromes/genetics , Mutation , Osteogenesis/genetics , Osteoporosis/genetics , Primary Immunodeficiency Diseases/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Humans
2.
Nat Commun ; 11(1): 3193, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32581213

ABSTRACT

Breast cancer is the most common type of cancer worldwide and one of the major causes of cancer death in women. Epidemiological studies have established a link between night-shift work and increased cancer risk, suggesting that circadian disruption may play a role in carcinogenesis. Here, we aim to shed light on the effect of chronic jetlag (JL) on mammary tumour development. To do this, we use a mouse model of spontaneous mammary tumourigenesis and subject it to chronic circadian disruption. We observe that circadian disruption significantly increases cancer-cell dissemination and lung metastasis. It also enhances the stemness and tumour-initiating potential of tumour cells and creates an immunosuppressive shift in the tumour microenvironment. Finally, our results suggest that the use of a CXCR2 inhibitor could correct the effect of JL on cancer-cell dissemination and metastasis. Altogether, our data provide a conceptual framework to better understand and manage the effects of chronic circadian disruption on breast cancer progression.


Subject(s)
Breast Neoplasms/pathology , Chronobiology Disorders/complications , Tumor Microenvironment/immunology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Cell Transformation, Neoplastic/drug effects , Chronic Disease , Chronobiology Disorders/genetics , Chronobiology Disorders/immunology , Cytokines/genetics , Female , Gene Expression Regulation , Immunosuppression Therapy , Light Signal Transduction/genetics , Mice , Mice, Transgenic , Neoplasm Metastasis/prevention & control , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/genetics
3.
Blood Adv ; 3(11): 1702-1713, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31171507

ABSTRACT

The bioactive lipid mediator sphingosine 1-phosphate (S1P) was recently assigned critical roles in platelet biology: whereas S1P1 receptor-mediated S1P gradient sensing was reported to be essential for directing proplatelet extensions from megakaryocytes (MKs) toward bone marrow sinusoids, MK sphingosine kinase 2 (Sphk2)-derived S1P was reported to further promote platelet shedding through receptor-independent intracellular actions, and platelet aggregation through S1P1 Yet clinical use of S1P pathway modulators including fingolimod has not been associated with risk of bleeding or thrombosis. We therefore revisited the role of S1P in platelet biology in mice. Surprisingly, no reduction in platelet counts was observed when the vascular S1P gradient was ablated by impairing S1P provision to plasma or S1P degradation in interstitial fluids, nor when gradient sensing was impaired by S1pr1 deletion selectively in MKs. Moreover, S1P1 expression and signaling were both undetectable in mature MKs in situ, and MK S1pr1 deletion did not affect platelet aggregation or spreading. When S1pr1 deletion was induced in hematopoietic progenitor cells, platelet counts were instead significantly elevated. Isolated global Sphk2 deficiency was associated with thrombocytopenia, but this was not replicated by MK-restricted Sphk2 deletion and was reversed by compound deletion of either Sphk1 or S1pr2, suggesting that this phenotype arises from increased S1P export and S1P2 activation secondary to redistribution of sphingosine to Sphk1. Consistent with clinical observations, we thus observe no essential role for S1P1 in facilitating platelet production or activation. Instead, S1P restricts megakaryopoiesis through S1P1, and can further suppress thrombopoiesis through S1P2 when aberrantly secreted in the hematopoietic niche.


Subject(s)
Blood Platelets/metabolism , Lysophospholipids/metabolism , Megakaryocytes/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Stem Cell Niche , Thrombopoiesis , Animals , Blood Platelets/cytology , Lysophospholipids/genetics , Megakaryocytes/cytology , Mice , Mice, Knockout , Sphingosine/genetics , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Sphingosine-1-Phosphate Receptors/metabolism
4.
J Bone Miner Res ; 34(2): 387-398, 2019 02.
Article in English | MEDLINE | ID: mdl-30347511

ABSTRACT

During skeletal mineralization, the sodium-phosphate co-transporter PiT1Slc20a1 is assumed to meet the phosphate requirements of bone-forming cells, although evidence is missing. Here, we used a conditional gene deletion approach to determine the role of PiT1 in growth plate chondrocytes. We show that PiT1 ablation shortly after birth generates a rapid and massive cell death in the center of the growth plate, together with an uncompensated endoplasmic reticulum (ER) stress, characterized by morphological changes and increased Chop, Atf4, and Bip expression. PiT1 expression in chondrocytes was not found at the cell membrane but co-localized with the ER marker ERp46, and was upregulated by the unfolded protein response cascade. In addition, we identified the protein disulfide isomerase (Pdi) ER chaperone as a PiT1 binding partner and showed that PiT1 ablation impaired Pdi reductase activity. The ER stress induced by PiT1 deficiency in chondrocytes was associated with intracellular retention of aggrecan and vascular endothelial growth factor A (Vegf-A), which was rescued by overexpressing a phosphate transport-deficient mutant of PiT1. Our data thus reveal a novel, Pi-transport independent function of PiT1, as a critical modulator of ER homeostasis and chondrocyte survival during endochondral ossification. © 2018 American Society for Bone and Mineral Research.


Subject(s)
Chondrocytes/metabolism , Endoplasmic Reticulum , Growth Plate/metabolism , Homeostasis , Osteogenesis , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Animals , Chondrocytes/cytology , Gene Expression Regulation , Growth Plate/cytology , Mice , Mice, Transgenic , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Unfolded Protein Response
5.
JCI Insight ; 3(17)2018 09 06.
Article in English | MEDLINE | ID: mdl-30185659

ABSTRACT

Sarcomas are still unsolved therapeutic challenges. Cancer stem cells are believed to contribute to sarcoma development, but lack of specific markers prevents their characterization and targeting. Here, we show that calpain-6 expression is associated with cancer stem cell features. In mouse models of bone sarcoma, calpain-6-expressing cells have unique tumor-initiating and metastatic capacities. Calpain-6 levels are especially high in tumors that have been successfully propagated in mouse to establish patient-derived xenografts. We found that calpain-6 levels are increased by hypoxia in vitro and calpain-6 is detected within hypoxic areas in tumors. Furthermore, calpain-6 expression depends on the stem cell transcription network that involves Oct4, Nanog, and Sox2 and is activated by hypoxia. Calpain-6 knockdown blocks tumor development in mouse and induces depletion of the cancer stem cell population. Data from transcriptomic analyses reveal that calpain-6 expression in sarcomas inversely correlates with senescence markers. Calpain-6 knockdown suppresses hypoxia-dependent prevention of senescence entry and also promotion of autophagic flux. Together, our results demonstrate that calpain-6 identifies sarcoma cells with stem-like properties and is a mediator of hypoxia to prevent senescence, promote autophagy, and maintain the tumor-initiating cell population. These findings open what we believe is a novel therapeutic avenue for targeting sarcoma stem cells.


Subject(s)
Autophagy , Calpain/metabolism , Cellular Senescence/physiology , Microtubule-Associated Proteins/metabolism , Neoplastic Stem Cells/metabolism , Sarcoma/metabolism , Animals , Biomarkers , Calpain/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , Disease Models, Animal , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Hypoxia , Male , Mice , Mice, Inbred BALB C , Microtubule-Associated Proteins/genetics , Nanog Homeobox Protein/metabolism , Neoplasms , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , Xenograft Model Antitumor Assays
6.
Oncoscience ; 5(5-6): 167-168, 2018 May.
Article in English | MEDLINE | ID: mdl-30035179
7.
Proc Natl Acad Sci U S A ; 115(5): E992-E1001, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29339479

ABSTRACT

Bone metastasis involves dynamic interplay between tumor cells and the local stromal environment. In bones, local hypoxia and activation of the hypoxia-inducible factor (HIF)-1α in osteoblasts are essential to maintain skeletal homeostasis. However, the role of osteoblast-specific HIF signaling in cancer metastasis is unknown. Here, we show that osteoprogenitor cells (OPCs) are located in hypoxic niches in the bone marrow and that activation of HIF signaling in these cells increases bone mass and favors breast cancer metastasis to bone locally. Remarkably, HIF signaling in osteoblast-lineage cells also promotes breast cancer growth and dissemination remotely, in the lungs and in other tissues distant from bones. Mechanistically, we found that activation of HIF signaling in OPCs increases blood levels of the chemokine C-X-C motif ligand 12 (CXCL12), which leads to a systemic increase of breast cancer cell proliferation and dissemination through direct activation of the CXCR4 receptor. Hence, our data reveal a previously unrecognized role of the hypoxic osteogenic niche in promoting tumorigenesis beyond the local bone microenvironment. They also support the concept that the skeleton is an important regulator of the systemic tumor environment.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Osteoblasts/metabolism , Alleles , Amino Acid Motifs , Animals , Bone Neoplasms/secondary , Bone and Bones/metabolism , Cell Lineage , Chemokine CXCL12/blood , Disease Progression , Female , Green Fluorescent Proteins/metabolism , Hypoxia , Ligands , Mice , Mice, Transgenic , Neoplasm Metastasis , Osteoclasts/metabolism , Signal Transduction
8.
Proc Natl Acad Sci U S A ; 113(19): 5453-8, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27122313

ABSTRACT

Low oxygen tension (hypoxia) regulates chondrocyte differentiation and metabolism. Hypoxia-inducible factor 1α (HIF1α) is a crucial hypoxic factor for chondrocyte growth and survival during development. The major metalloproteinase matrix metalloproteinase 13 (MMP13) is also associated with chondrocyte hypertrophy in adult articular cartilage, the lack of which protects from cartilage degradation and osteoarthritis (OA) in mice. MMP13 is up-regulated by the Wnt/ß-catenin signaling, a pathway involved in chondrocyte catabolism and OA. We studied the role of HIF1α in regulating Wnt signaling in cartilage and OA. We used mice with conditional knockout of Hif1α (∆Hif1α(chon)) with joint instability. Specific loss of HIF1α exacerbated MMP13 expression and cartilage destruction. Analysis of Wnt signaling in hypoxic chondrocytes showed that HIF1α lowered transcription factor 4 (TCF4)-ß-catenin transcriptional activity and inhibited MMP13 expression. Indeed, HIF1α interacting with ß-catenin displaced TCF4 from MMP13 regulatory sequences. Finally, ΔHif1α(chon) mice with OA that were injected intraarticularly with PKF118-310, an inhibitor of TCF4-ß-catenin interaction, showed less cartilage degradation and reduced MMP13 expression in cartilage. Therefore, HIF1α-ß-catenin interaction is a negative regulator of Wnt signaling and MMP13 transcription, thus reducing catabolism in OA. Our study contributes to the understanding of the role of HIF1α in OA and highlights the HIF1α-ß-catenin interaction, thus providing new insights into the impact of hypoxia in articular cartilage.


Subject(s)
Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Matrix Metalloproteinase 13/metabolism , Osteoarthritis/metabolism , beta Catenin/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Osteoarthritis/pathology , Protein Binding , Signal Transduction
9.
Med Sci (Paris) ; 30(4): 366-71, 2014 Apr.
Article in French | MEDLINE | ID: mdl-24801029

ABSTRACT

In spite of a good understanding of the cellular and molecular mechanisms responsible for neoplastic transformation, most cancer therapies remain partially effective and transient. Recent studies established the importance of the tumor microenvironment in tumor growth and dissemination, and in the resistance to antitumor therapies. This review summarizes our current knowledge of the local and systemic effects, sometimes surprising, which result from interactions between neoplastic cancer cells and their microenvironment.


Subject(s)
Neoplasm Metastasis/pathology , Neoplasms/pathology , Tumor Microenvironment , Humans , Tumor Cells, Cultured
10.
Nat Cell Biol ; 15(2): 201-13, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23354167

ABSTRACT

Despite advances in our understanding of breast cancer, patients with metastatic disease have poor prognoses. GATA3 is a transcription factor that specifies and maintains mammary luminal epithelial cell fate, and its expression is lost in breast cancer, correlating with a worse prognosis in human patients. Here, we show that GATA3 promotes differentiation, suppresses metastasis and alters the tumour microenvironment in breast cancer by inducing microRNA-29b (miR-29b) expression. Accordingly, miR-29b is enriched in luminal breast cancers and loss of miR-29b, even in GATA3-expressing cells, increases metastasis and promotes a mesenchymal phenotype. Mechanistically, miR-29b inhibits metastasis by targeting a network of pro-metastatic regulators involved in angiogenesis, collagen remodelling and proteolysis, including VEGFA, ANGPTL4, PDGF, LOX and MMP9, and targeting ITGA6, ITGB1 and TGFB, thereby indirectly affecting differentiation and epithelial plasticity. The discovery that a GATA3-miR-29b axis regulates the tumour microenvironment and inhibits metastasis opens up possibilities for therapeutic intervention in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , GATA3 Transcription Factor/metabolism , MicroRNAs/metabolism , Tumor Microenvironment , Animals , Breast Neoplasms/genetics , Breast Neoplasms/secondary , Cell Differentiation , Cell Line, Tumor , Female , GATA3 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genotype , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Phenotype , Transfection , Up-Regulation
11.
J Bone Miner Res ; 25(11): 2504-14, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20533303

ABSTRACT

Wdr5, a bone morphogenetic protein 2 (BMP-2)-induced protein belonging to the family of the WD repeat proteins, is expressed in proliferating and hypertrophic chondrocytes of the growth plate and in osteoblasts. Although previous studies have provided insight into the mechanisms by which Wdr5 affects chondrocyte and osteoblast differentiation, whether Wdr5 is required in vivo for endochondral bone development has not been addressed. In this study, using an avian replication competent retrovirus (RCAS) system delivering Wdr5 short hairpin (sh) RNA to silence Wdr5 in the developing limb, we report that reduction of Wdr5 levels delays endochondral bone development and consequently results in shortening of the skeletal elements. Shortening of the skeletal elements was due to impaired chondrocyte maturation, evidenced by a significant reduction of Runx2, type X collagen, and osteopontin expression. A decrease in Runx2, type collagen I, and ostepontin expression in osteoblasts and a subsequent defect in mineralized bone was observed as well when Wdr5 levels were reduced. Most important, retroviral misexpression of Runx2 rescued the phenotype induced by Wdr5 shRNA. These findings suggest that during limb development, Wdr5 is required for endochondral bone formation and that Wdr5 influences this process, at least in part, by regulating Runx2 expression.


Subject(s)
Avian Proteins/metabolism , Bone Development , Chickens/growth & development , Animals , Avian Proteins/genetics , Bone Development/genetics , Cell Differentiation/genetics , Chick Embryo , Chondrocytes/metabolism , Chondrocytes/pathology , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation, Developmental , Hypertrophy , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/genetics , Phenotype , RNA, Small Interfering/metabolism , Retroviridae/genetics
12.
J Cell Physiol ; 222(1): 42-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19798694

ABSTRACT

There is increasing evidence that the numerous mechanisms that regulate cell differentiation during normal development are also involved in tumorigenesis. In breast cancer, differentiation markers expressed by the primary tumor are routinely profiled to guide clinical decisions. Indeed, numerous studies have shown that the differentiation profile correlates with the metastatic potential of tumors. The transcription factor GATA3 has emerged recently as a strong predictor of clinical outcome in human luminal breast cancer. In the mammary gland, GATA3 is required for luminal epithelial cell differentiation and commitment, and its expression is progressively lost during luminal breast cancer progression as cancer cells acquire a stem cell-like phenotype. Importantly, expression of GATA3 in GATA3-negative, undifferentiated breast carcinoma cells is sufficient to induce tumor differentiation and inhibits tumor dissemination in a mouse model. These findings demonstrate the exquisite ability of a differentiation factor to affect malignant properties, and raise the possibility that GATA3 or its downstream genes could be used in treating luminal breast cancer. This review highlights our recent understanding of GATA3 in both normal mammary development and tumor differentiation.


Subject(s)
Breast Neoplasms/pathology , Cell Differentiation , Embryonic Development , GATA3 Transcription Factor/metabolism , Animals , Female , Humans , Neoplasm Metastasis/pathology , Organ Specificity
13.
Mol Cell Biol ; 28(1): 344-57, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17967876

ABSTRACT

Parathyroid hormone-related peptide (PTHrP) and the parathyroid hormone-PTHrP receptor increase chondrocyte proliferation and delay chondrocyte maturation in endochondral bone development at least partly through cyclic AMP (cAMP)-dependent signaling pathways. Because data suggest that the ability of cAMP to stimulate cell proliferation involves the mitogen-activated protein kinase kinase kinase B-Raf, we hypothesized that B-Raf might mediate the proliferative action of PTHrP in chondrocytes. Though B-Raf is expressed in proliferative chondrocytes, its conditional removal from cartilage did not affect chondrocyte proliferation and maturation or PTHrP-induced chondrocyte proliferation and PTHrP-delayed maturation. Similar results were obtained by conditionally removing B-Raf from osteoblasts. Because A-raf and B-raf are expressed similarly in cartilage, we speculated that they may fulfill redundant functions in this tissue. Surprisingly, mice with chondrocytes deficient in both A-Raf and B-Raf exhibited normal endochondral bone development. Activated extracellular signal-regulated kinase (ERK) was detected primarily in hypertrophic chondrocytes, where C-raf is expressed, and the suppression of ERK activation in these cells by PTHrP or a MEK inhibitor coincided with a delay in chondrocyte maturation. Taken together, these results demonstrate that B-Raf and A-Raf are dispensable for endochondral bone development and they indicate that the main role of ERK in cartilage is to stimulate not cell proliferation, but rather chondrocyte maturation.


Subject(s)
Bone and Bones/metabolism , Chondrocytes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypertrophy/metabolism , Parathyroid Hormone-Related Protein/metabolism , Proto-Oncogene Proteins A-raf/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Animals , Cartilage/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Enzyme Activation , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteogenesis , Parathyroid Hormone-Related Protein/genetics , Proto-Oncogene Proteins A-raf/deficiency , Proto-Oncogene Proteins A-raf/genetics , Proto-Oncogene Proteins B-raf/deficiency , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction
14.
Ann N Y Acad Sci ; 1117: 26-39, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18056035

ABSTRACT

Fetal growth plate chondrocyte is a unique mesenchymal tissue, as it is avascular and hypoxic. Yet, chondrocytes not only survive in this environment, but also undergo all cellular processes (proliferation, growth arrest, differentiation, etc.) required for normal endochondral bone development. A crucial mediator of the adaptive response of cells to hypoxia is a transcription factor named hypoxia-inducible factor 1alpha (Hif-1alpha). One target of Hif-1alpha transcriptional activation is the angiogenic factor vascular endothelial growth factor (VEGF), whereas Hif-1alpha accumulation is controlled by the von Hippel-Lindau (VHL) tumor suppressor, an E3-ubiquitin ligase that induces its degradation by the proteasome. We, and others, demonstrated that each component of this pathway is a critical regulator of endochondral bone development. In particular, we previously established that Hif-1alpha is a survival factor for hypoxic chondrocytes, and that it also negatively regulates cell proliferation. Interestingly, we also showed that hypoxia increases extracellular matrix accumulation in a Hif-1alpha-dependent fashion. This suggested that Hif-1alpha could be critically important not only for cell survival and proliferation but also for cell differentiation. We recently demonstrated that Hif-1alpha is indeed a differentiation factor since it is required in mesenchymal cells both for early chondrogenesis, and for joint development.


Subject(s)
Hypoxia , Animals , Bone Development , Bone and Bones/metabolism , Cell Differentiation , Cell Proliferation , Cell Survival , Chondrocytes/metabolism , Developmental Biology/methods , Growth Plate/embryology , Humans , Mesoderm/metabolism , Models, Biological , Proteasome Endopeptidase Complex/metabolism , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
15.
J Cell Biol ; 177(3): 451-64, 2007 May 07.
Article in English | MEDLINE | ID: mdl-17470636

ABSTRACT

Recent evidence suggests that low oxygen tension (hypoxia) may control fetal development and differentiation. A crucial mediator of the adaptive response of cells to hypoxia is the transcription factor Hif-1alpha. In this study, we provide evidence that mesenchymal condensations that give origin to endochondral bones are hypoxic during fetal development, and we demonstrate that Hif-1alpha is expressed and transcriptionally active in limb bud mesenchyme and in mesenchymal condensations. To investigate the role of Hif-1alpha in mesenchymal condensations and in early chondrogenesis, we conditionally inactivated Hif-1alpha in limb bud mesenchyme using a Prx1 promoter-driven Cre transgenic mouse. Conditional knockout of Hif-1alpha in limb bud mesenchyme does not impair mesenchyme condensation, but alters the formation of the cartilaginous primordia. Late hypertrophic differentiation is also affected as a result of the delay in early chondrogenesis. In addition, mutant mice show a striking impairment of joint development. Our study demonstrates a crucial, and previously unrecognized, role of Hif-1alpha in early chondrogenesis and joint formation.


Subject(s)
Chondrogenesis/physiology , Hindlimb/embryology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Joints/embryology , Mesoderm/metabolism , Animals , Cell Hypoxia/physiology , Female , Hindlimb/cytology , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Joints/cytology , Mesoderm/cytology , Mice , Mice, Knockout , Pregnancy
16.
Development ; 133(4): 651-62, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16421188

ABSTRACT

Parathyroid hormone-related protein (PTHrP) is essential to maintain a pool of dividing, immature chondrocytes in the growth plate of long bones. In chick and mouse, expression of Nkx3.2/Bapx1 in the growth plate is restricted to the proliferative zone and is down regulated as chondrocyte maturation begins. Nkx3.2/Bapx1 expression is lost in the growth plates of mice engineered to lack PTHrP signaling and, conversely, is maintained by ectopic expression of PTHrP in developing bones. Artificially preventing Nkx3.2/Bapx1 downregulation, by forced expression of either retroviral-encoded PTHrP or Nkx3.2 inhibits chondrocyte maturation. Although wild-type Nkx3.2 blocks chondrocyte maturation by acting as a transcriptional repressor, a ;reverse function' mutant of Nkx3.2 that has been converted into a transcriptional activator conversely accelerates chondrocyte maturation. Nkx3.2 represses expression of the chondrocyte maturation factor Runx2, and Runx2 mis-expression can rescue the Nkx3.2-induced blockade of chondrocyte maturation. Taken together, these results suggest that PTHrP signals block chondrocyte hypertrophy by, in part, maintaining the expression of Nkx3.2/Bapx1, which in turn represses the expression of genes required for chondrocyte maturation.


Subject(s)
Cell Proliferation , Chondrocytes/cytology , Growth Plate/cytology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Chick Embryo , Chondrocytes/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation, Developmental , Growth Plate/metabolism , Homeodomain Proteins/genetics , Mice , Parathyroid Hormone-Related Protein/physiology , Signal Transduction , Transcription Factors/genetics
17.
Biochem Biophys Res Commun ; 328(3): 658-65, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15694399

ABSTRACT

Endochondral bone development is a complex process in which undifferentiated mesenchymal cells differentiate into chondrocytes, which then undergo well-ordered and controlled phases of proliferation, hypertrophic differentiation, death, blood vessel invasion, and finally replacement of cartilage with bone. The process recapitulates basic and fundamental mechanisms of cell biology with a highly specific spatial and temporal pattern, and it thus constitutes an excellent model for the analysis of such mechanisms. In recent years, the tools provided by modern genetic both in mice and men have been instrumental in the process of identifying and dissecting basic molecular mechanisms of endochondral bone formation. This review is a brief summary of the current knowledge about some of the crucial factors involved in growth plate development.


Subject(s)
Bone Development/physiology , Bone and Bones/embryology , Bone and Bones/physiology , Chondrocytes/physiology , Gene Expression Regulation, Developmental/physiology , Osteocytes/physiology , Transcription Factors/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Chondrocytes/cytology , Chondrogenesis/physiology , Hedgehog Proteins , Humans , Mice , Neoplasm Proteins/metabolism , Osteocytes/cytology , Parathyroid Hormone-Related Protein/metabolism , Trans-Activators/metabolism
18.
Birth Defects Res C Embryo Today ; 69(4): 352-62, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14745975

ABSTRACT

Endochondral bone development is a fascinating story of proliferation, maturation, and death. An understanding of this process at the molecular level is emerging. In particular, significant advances have been made in understanding the role of parathyroid-hormone-related peptide (PTHrP), parathyroid hormone (PTH), and the PTH/PTHrP receptor in endochondral bone development. Mutations of the PTH/PTHrP receptor have been identified in Jansen metaphyseal chondrodysplasia, Blomstrand's lethal chondrodysplasia, and enchondromatosis. Furthermore, genetic manipulations of the PTHrP, PTH, and the PTH/PTHrP receptor genes, respectively, have demonstrated the critical role of these proteins in regulating both the switch between proliferation and differentiation of chondrocytes, and their replacement by bone cells. A future area of investigation will be the identification of downstream effectors of PTH, PTHrP, and PTH/PTHrP receptor activities. Furthermore, it will be of critical importance to study how these proteins cooperate and integrate with other molecules that are essential for growth plate development.


Subject(s)
Bone Development , Parathyroid Hormone-Related Protein/physiology , Parathyroid Hormone/physiology , Receptor, Parathyroid Hormone, Type 1/physiology , Animals , Cell Differentiation , Chondrocytes/physiology , Dwarfism/genetics , Enchondromatosis/genetics , Female , Growth Plate/physiology , Humans , Male , Mice , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Parathyroid Hormone/genetics , Parathyroid Hormone-Related Protein/genetics , Receptor, Parathyroid Hormone, Type 1/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...